Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Barry C. Jones is active.

Publication


Featured researches published by Barry C. Jones.


Drug Metabolism and Disposition | 2004

DRUG-DRUG INTERACTIONS FOR UDP-GLUCURONOSYLTRANSFERASE SUBSTRATES: A PHARMACOKINETIC EXPLANATION FOR TYPICALLY OBSERVED LOW EXPOSURE (AUCI/AUC) RATIOS

J. Andrew Williams; Ruth Hyland; Barry C. Jones; Dennis A. Smith; Susan Hurst; Theunis C. Goosen; Vincent Peterkin; Jeffrey R. Koup; Simon E. Ball

Glucuronidation is a listed clearance mechanism for 1 in 10 of the top 200 prescribed drugs. The objective of this article is to encourage those studying ligand interactions with UDP-glucuronosyltransferases (UGTs) to adequately consider the potential consequences of in vitro UGT inhibition in humans. Spurred on by interest in developing potent and selective inhibitors for improved confidence around UGT reaction phenotyping, and the increased availability of recombinant forms of human UGTs, several recent studies have reported in vitro inhibition of UGT enzymes. In some cases, the observed potency of UGT inhibitors in vitro has been interpreted as having potential relevance in humans via pharmacokinetic drug-drug interactions. Although there are reported examples of clinically relevant drug-drug interactions for UGT substrates, exposure increases of the aglycone are rarely greater than 100% in the presence of an inhibitor relative to its absence (i.e., AUCi/AUC ≤2). This small magnitude in change is in contrast to drugs primarily cleared by cytochrome P450 enzymes, where exposures have been reported to increase as much as 35-fold on coadministration with an inhibitor (e.g., ketoconazole inhibition of CYP3A4-catalyzed terfenadine metabolism). In this article the evidence for purported clinical relevance of potent in vitro inhibition of UGT enzymes will be assessed, taking the following into account: in vitro data on the enzymology of glucuronide formation from aglycone, pharmacokinetic principles based on empirical data for inhibition of metabolism, and clinical data on the pharmacokinetic drug-drug interactions of drugs primarily cleared by glucuronidation.


Biochemical Pharmacology | 1992

Speculations on the Substrate Structure-Activity Relationship (SSAR) of cytochrome P450 enzymes

Dennis A. Smith; Barry C. Jones

This brief review attempts to define the SSAR of two families of cytochrome P450. With P4502D catalytic competence is achieved by tight ionic binding which gives the enzyme high regioselectivity. In contrast P4503A achieves catalytic competence by a flexible binding site relying on hydrophobic forces that allow chemically vulnerable sites to be the principal sites of metabolism. In general, the different binding mechanism should be reflected in the enzyme, such that substrates of P4502D should have lower Km values than substrates of P4503A. Thus, routes of metabolism catalysed by P4502D may be saturated at substrate concentrations lower than routes catalysed by P4503A. The apparent differences between P4502D and P4503A in terms of substrate specificity bring into question what relationships govern other families of cytochrome P450. Our analysis of data suggests that the other principal form involved, generally, in the metabolism of pharmaceuticals in humans is P4502C9 (possibly 2C8 and 2C10). The enzyme is responsible for the metabolism of phenytoin, tolbutamide, tienilic acid [4], naproxen, ibuprofen, diclofenac [38], the 7-hydroxylation of S-warfarin [39] and the 7-hydroxylation of delta 1-tetrahydrocannabinol [40]. These compounds all have areas of strong hydrogen bond [4] forming potential (Fig. 8), all distanced 5-10A from the site of metabolism. Moreover the carboxylic acid function of naproxen, ibuprofen and diclofenac (pKa 4.5) and the sulfonylurea of tolbutamide (pKa 5.4) render the compounds ionized at physiological pH. The ionised group is positioned 7-11A from the site of metabolism. It is likely, therefore, that hydrogen bonding and possibly ion-pair interactions play a major role in determining the SSAR of the P4502C isoenzymes. These interactions would suggest that the P4502C enzymes are analogous to P4502D rather than P4503A. In this regard it is noteworthy that P4502C9 is selectively and potently inhibited by sulfaphenazole (IC50 of 0.6 microM), a compound that is structurally related (Fig. 8) to the substrates in terms of potential hydrogen bonding regions [4, 41]. Simplistically we suggest that the SSAR of the various P450 enzymes ranges from the highly selective enzymes dealing with endogenous substrates, through the enzymes metabolising exogenous substrates with narrow substrate structure requirements such as P4502D to P4503A with its broad substrate structure range. It would seem logical that animals and humans would evolve such combinations of isoenzymes to deal with the vast array of exogenous xenobiotics.


Journal of Computer-aided Molecular Design | 2001

Lipophilicity in PK design: methyl, ethyl, futile.

Han van de Waterbeemd; Dennis A. Smith; Barry C. Jones

Lipophilicity, often expressed as distribution coefficients (log D) in octanol/water, is an important physicochemical parameter influencing processes such as oral absorption, brain uptake and various pharmacokinetic (PK) properties. Increasing log D values increases oral absorption, plasma protein binding and volume of distribution. However, more lipophilic compounds also become more vulnerable to P450 metabolism, leading to higher clearance. Molecular size and hydrogen bonding capacity are two other properties often considered as important for membrane permeation and pharmacokinetics. Interrelationships among these physicochemical properties are discussed. Increasing size (molecular weight) often gives higher potency, but inevitably also leads to either higher lipophilicity, and hence poorer dissolution/solubility, or to more hydrogen bonding capacity, which limits oral absorption. Differences in optimal properties between gastrointestinal absorption and uptake into the brain are addressed. Special attention is given to the desired lipophilicity of CNS drugs. In examples using β-blockers, Ca channel antagonists and peptidic renin inhibitors we will demonstrate how potency and pharmacokinetic properties need to be balanced.


Medicinal Research Reviews | 1996

Design of drugs involving the concepts and theories of drug metabolism and pharmacokinetics.

Dennis A. Smith; Barry C. Jones; D. K. Walker

Drug metabolism input to the discovery process had historically been on an empirical case-by-case basis, since, detailed descriptors of the effect on pharmacokinetics of a change in structure or physicochemical property were not available. Considerable advances have been made in recent years, such that basic rules can be applied to predict the behavior of a compound in man based on physicochemistry and structure. This is particularly true in the areas of absorption, distribution, and clearance. In particular, knowledge of the reactions catalyzed by the enzymes of drug metabolism, including the cytochrome P450 super family, can be used in the design of new chemical entities, together with the usual pharmacological-derived SAR. The combination of both pharmacokinetics and pharmacodynamics at the discovery stage leads to drugs with optimum performance characteristics. Such drugs are easier to develop, representing a huge saving in resources. Moreover, the marketed compound is much more likely to find high clinical utilization. This review uses dofetilide, fluconazole, and amlodipine to highlight the multifaceted consequences of changing chemical structure, in terms of drug disposition, and reinforces these principles with examples from the literature.


Drug Discovery Today | 1997

Properties of cytochrome P450 isoenzymes and their substrates part 2: properties of cytochrome P450 substrates

Dennis A. Smith; Mark J. Ackland; Barry C. Jones

Cytochrome P450 isoenzymes are pivotal in drug clearance. Part 1 of this two-part review, published in the October issue of Drug Discovery Today , described the active site characteristics of members of the P450 superfamily. This article describes the great increase in our understanding of the substrate requirements of the human cytochrome P450 family and highlights the relevance of this knowledge for the design of new therapeutic agents.


Xenobiotica | 1998

Human cytochrome P450s: selectivity and measurement in vivo

Dennis A. Smith; S. Abel; Ruth Hyland; Barry C. Jones

This review considers the human cytochrome P450 (CYP) enzymes responsible for the metabolism of drugs. This area has generated considerable interest over the past 10 or so years due to the identi® cation of the major human isoforms and some rationalization of the reasons for their selectivity towards various substrate classes (Smith and Jones 1992). At the same time, the pharmacokinetic behaviour of these same substrates has been investigated and intimately linked to the relative in vivo expression of each isoform. Thus a new chemical structure can be rapidly assessed as to which isoform(s) is important in its metabolism and then predictions made as to its likely in vivo performance particularly with regard to its variability. The knowledge has come from studying the pharmacokinetics of compounds which are metabolized by speci® c isoforms. Historically, the in vivo formation has been generated before the isozyme responsible for metabolism has been identi® ed, although such examples will in future became an increasing rarity. It is the purpose of this review to cover developments in these two intimately linked areas: why a compound is metabolized by a particular isoform (substrate± structure activity relationships, SSAR) and how we assess the expression of that isoform in vivo.


British Journal of Clinical Pharmacology | 2008

Maraviroc: in vitro assessment of drug–drug interaction potential

Ruth Hyland; Maurice Dickins; Claire Collins; Hannah M. Jones; Barry C. Jones

AIMS To characterize the cytochrome P450 enzyme(s) responsible for the N-dealkylation of maraviroc in vitro, and predict the extent of clinical drug-drug interactions (DDIs). METHODS Human liver and recombinant CYP microsomes were used to identify the CYP enzyme responsible for maraviroc N-dealkylation. Studies comprised enzyme kinetics and evaluation of the effects of specific CYP inhibitors. In vitro data were then used as inputs for simulation of DDIs with ketoconazole, ritonavir, saquinavir and atazanvir, using the Simcyptrade mark population-based absorption, distribution, metabolism and elimination (ADME) simulator. Study designs for simulations mirrored those actually used in the clinic. RESULTS Maraviroc was metabolized to its N-dealkylated product via a single CYP enzyme characterized by a K(m) of 21 microM and V(max) of 0.45 pmol pmol(-1) min(-1) in human liver microsomes and was inhibited by ketoconazole (CYP3A4 inhibitor). In a panel of recombinant CYP enzymes, CYP3A4 was identified as the major CYP responsible for maraviroc metabolism. Using recombinant CYP3A4, N-dealkylation was characterized by a K(m) of 13 microM and a V(max) of 3 pmol pmol(-1) CYP min(-1). Simulations therefore focused on the effect of CYP3A4 inhibitors on maraviroc pharmacokinetics. The simulated median AUC ratios were in good agreement with observed clinical changes (within twofold in all cases), although, in general, there was a trend for overprediction in the magnitude of the DDI. CONCLUSION Maraviroc is a substrate for CYP3A4, and exposure will therefore be modulated by CYP3A4 inhibitors. Simcyptrade mark has successfully simulated the extent of clinical interactions with CYP3A4 inhibitors, further validating this software as a good predictor of CYP-based DDIs.


Drug Discovery Today | 1997

Properties of cytochrome P450 isoenzymes and their substrates Part 1: active site characteristics

Dennis A. Smith; Mark J. Ackland; Barry C. Jones

Cytochrome P450 isoenzymes are pivotal in drug clearance. Protein homology modelling combined with studies on the structure and physico-chemistry of substrates is allowing the key factors governing selectivity for substrates to be ascertained. The knowledge gained will be used in future in the drug design process to produce compounds more resistant to metabolism or with lowered potential to inhibit the enzyme. Ultimately, therefore, the progress being made will result in the discovery and development of safer and more effective therapies.


Clinical Pharmacokinectics | 2011

Simulation of Human Intravenous and Oral Pharmacokinetics of 21 Diverse Compounds Using Physiologically Based Pharmacokinetic Modelling

Hannah M. Jones; Iain Gardner; Wendy Collard; Phil Stanley; Penny Oxley; Natilie Hosea; David R. Plowchalk; Steve S. Gernhardt; Jing Lin; Maurice Dickins; S. Ravi Rahavendran; Barry C. Jones; Kenny J. Watson; Henry Pertinez; Vikas Kumar; Susan Cole

AbstractBackground: The importance of predicting human pharmacokinetics during compound selection has been recognized in the pharmaceutical industry. To this end there are many different approaches that are applied. Methods: In this study we compared the accuracy of physiologically based pharmacokinetic (PBPK) methodologies implemented in GastroPlus™ with the one-compartment approach routinely used at Pfizer for human pharmacokinetic plasma concentration-time profile prediction. Twenty-one Pfizer compounds were selected based on the availability of relevant preclinical and clinical data. Intravenous and oral human simulations were performed for each compound. To understand any mispredictions, simulations were also performed using the observed clearance (CL) value as input into the model. Results: The simulation results using PBPK were shown to be superior to those obtained via traditional one-compartment analyses. In many cases, this difference was statistically significant. Specifically, the results showed that the PBPK approach was able to accurately predict passive distribution and absorption processes. Some issues and limitations remain with respect to the prediction of CL and active transport processes and these need to be improved to further increase the utility of PBPK modelling. A particular advantage of the PBPK approach is its ability to accurately predict the multiphasic shape of the pharmacokinetic profiles for many of the compounds tested. Conclusion: The results from this evaluation demonstrate the utility of PBPK methodology for the prediction of human pharmacokinetics. This methodology can be applied at different stages to enhance the understanding of the compounds in a particular chemical series, guide experiments, aid candidate selection and inform clinical trial design.


Xenobiotica | 2012

Application of PBPK modelling in drug discovery and development at Pfizer

Hannah M. Jones; Maurice Dickins; Kuresh Youdim; James R. Gosset; Neil Attkins; Tanya L. Hay; Ian Gurrell; Y. Raj Logan; Peter J. Bungay; Barry C. Jones; Iain Gardner

Early prediction of human pharmacokinetics (PK) and drug–drug interactions (DDI) in drug discovery and development allows for more informed decision making. Physiologically based pharmacokinetic (PBPK) modelling can be used to answer a number of questions throughout the process of drug discovery and development and is thus becoming a very popular tool. PBPK models provide the opportunity to integrate key input parameters from different sources to not only estimate PK parameters and plasma concentration-time profiles, but also to gain mechanistic insight into compound properties. Using examples from the literature and our own company, we have shown how PBPK techniques can be utilized through the stages of drug discovery and development to increase efficiency, reduce the need for animal studies, replace clinical trials and to increase PK understanding. Given the mechanistic nature of these models, the future use of PBPK modelling in drug discovery and development is promising, however, some limitations need to be addressed to realize its application and utility more broadly.

Researchain Logo
Decentralizing Knowledge